Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Glutamine independence is a selectable feature of pluripotent stem cells

Abstract

Most rapidly proliferating mammalian cells rely on the oxidation of exogenous glutamine to support cell proliferation. We previously found that culture of mouse embryonic stem cells in the presence of inhibitors against mitogen-activated protein kinase kinase and glycogen synthase kinase 3 beta to maintain pluripotency reduces cellular reliance on glutamine for tricarboxylic acid cycle anaplerosis, enabling embryonic stem cells to proliferate in the absence of exogenous glutamine. Here we show that reduced dependence on exogenous glutamine is a generalizable feature of pluripotent stem cells. Enhancing self-renewal, through either overexpression of pluripotency-associated transcription factors or altered signal transduction, decreases the use of glutamine-derived carbons in the tricarboxylic acid cycle. As a result, cells with the highest potential for self-renewal can be enriched by transient culture in glutamine-deficient media. During pluripotent cell culture or reprogramming to pluripotency, transient glutamine withdrawal selectively leads to the elimination of non-pluripotent cells. These data reveal that reduced dependence on glutamine anaplerosis is an inherent feature of self-renewing pluripotent stem cells and reveal a simple, non-invasive mechanism to select for mouse and human pluripotent stem cells within a heterogeneous population during both embryonic stem cell passage and induced pluripotent cell reprogramming.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Glutamine anaplerosis is reduced in ESCs with enhanced self-renewal.
Fig. 2: Enhanced self-renewal improves glutamine-independent survival.
Fig. 3: Transient glutamine withdrawal enhances ESC self-renewal.
Fig. 4: Transient glutamine withdrawal improves mouse somatic cell reprogramming to pluripotency and enhances human ESC self-renewal.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request. Source data for all gas-chromatography–mass spectrometry data are provided in Supplementary Table 1.

Code availability

The MATLAB code that supports the findings of this study is also available from the corresponding author upon reasonable request.

References

  1. DeBerardinis, R. J., Lum, J. J., Hatzivassiliou, G. & Thompson, C. B. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 7, 11–20 (2008).

    Article  CAS  Google Scholar 

  2. Palm, W. & Thompson, C. B. Nutrient acquisition strategies of mammalian cells. Nature 546, 234–242 (2017).

    Article  CAS  Google Scholar 

  3. Vander Heiden, M. G. & DeBerardinis, R. J. Understanding the intersections between metabolism and cancer biology. Cell 168, 657–669 (2017).

    Article  CAS  Google Scholar 

  4. Schvartzman, J. M., Thompson, C. B. & Finley, L. W. S. Metabolic regulation of chromatin modifications and gene expression. J. Cell Biol. 217, 2247–2259 (2018).

    Article  CAS  Google Scholar 

  5. Saxton, R. A. & Sabatini, D. M. mTOR signaling in growth, metabolism, and disease. Cell 168, 960–976 (2017).

    Article  CAS  Google Scholar 

  6. Su, X., Wellen, K. E. & Rabinowitz, J. D. Metabolic control of methylation and acetylation. Curr. Opin. Chem. Biol. 30, 52–60 (2016).

    Article  CAS  Google Scholar 

  7. Lu, C. & Thompson, C. B. Metabolic regulation of epigenetics. Cell Metab. 16, 9–17 (2012).

    Article  CAS  Google Scholar 

  8. Carey, B. W., Finley, L. W., Cross, J. R., Allis, C. D. & Thompson, C. B. Intracellular α-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature 518, 413–416 (2015).

    Article  CAS  Google Scholar 

  9. Gu, W. et al. Glycolytic metabolism plays a functional role in regulating human pluripotent stem cell state. Cell Stem Cell 19, 476–490 (2016).

    Article  CAS  Google Scholar 

  10. Tohyama, S. et al. Glutamine oxidation is indispensable for survival of human pluripotent stem cells. Cell Metab. 23, 663–674 (2016).

    Article  CAS  Google Scholar 

  11. Zhang, H. et al. Distinct metabolic states can support self-renewal and lipogenesis in human pluripotent stem cells under different culture conditions. Cell Rep. 16, 1536–1547 (2016).

    Article  CAS  Google Scholar 

  12. Hwang, I. Y. et al. Psat1-dependent fluctuations in α-ketoglutarate affect the timing of ESC differentiation. Cell Metab. 24, 494–501 (2016).

    Article  CAS  Google Scholar 

  13. Moussaieff, A. et al. Glycolysis-mediated changes in acetyl-CoA and histone acetylation control the early differentiation of embryonic stem cells. Cell Metab. 21, 392–402 (2015).

    Article  CAS  Google Scholar 

  14. TeSlaa, T. et al. α-Ketoglutarate accelerates the initial differentiation of primed human pluripotent stem cells. Cell Metab. 24, 485–493 (2016).

    Article  CAS  Google Scholar 

  15. Chambers, I. et al. Nanog safeguards pluripotency and mediates germline development. Nature 450, 1230–1234 (2007).

    Article  CAS  Google Scholar 

  16. Filipczyk, A. et al. Biallelic expression of nanog protein in mouse embryonic stem cells. Cell Stem Cell 13, 12–13 (2013).

    Article  CAS  Google Scholar 

  17. Ying, Q. L. et al. The ground state of embryonic stem cell self-renewal. Nature 453, 519–523 (2008).

    Article  CAS  Google Scholar 

  18. Chisolm, D. A. et al. CCCTC-binding factor translates interleukin 2- and α-ketoglutarate-sensitive metabolic changes in T cells into context-dependent gene programs. Immunity 47, 251–267.e7 (2017).

    Article  CAS  Google Scholar 

  19. Liu, P. S. et al. α-Ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat. Immunol. 18, 985–994 (2017).

    Article  CAS  Google Scholar 

  20. Yang, Q. et al. AMPK/α-ketoglutarate axis dynamically mediates DNA demethylation in the Prdm16 promoter and brown adipogenesis. Cell Metab. 24, 542–554 (2016).

    Article  CAS  Google Scholar 

  21. Burdon, T., Stracey, C., Chambers, I., Nichols, J. & Smith, A. Suppression of SHP-2 and ERK signalling promotes self-renewal of mouse embryonic stem cells. Dev. Biol. 210, 30–43 (1999).

    Article  CAS  Google Scholar 

  22. van Oosten, A. L., Costa, Y., Smith, A. & Silva, J. C. JAK/STAT3 signalling is sufficient and dominant over antagonistic cues for the establishment of naive pluripotency. Nat. Commun. 3, 817 (2012).

    Article  Google Scholar 

  23. Martello, G., Bertone, P. & Smith, A. Identification of the missing pluripotency mediator downstream of leukaemia inhibitory factor. EMBO J. 32, 2561–2574 (2013).

    Article  CAS  Google Scholar 

  24. Chambers, I. et al. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell 113, 643–655 (2003).

    Article  CAS  Google Scholar 

  25. Mitsui, K. et al. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell 113, 631–642 (2003).

    Article  CAS  Google Scholar 

  26. Zhang, P., Andrianakos, R., Yang, Y., Liu, C. & Lu, W. Kruppel-like factor 4 (Klf4) prevents embryonic stem (ES) cell differentiation by regulating Nanog gene expression. J. Biol. Chem. 285, 9180–9189 (2010).

    Article  CAS  Google Scholar 

  27. Festuccia, N. et al. Esrrb is a direct Nanog target gene that can substitute for Nanog function in pluripotent cells. Cell Stem Cell 11, 477–490 (2012).

    Article  CAS  Google Scholar 

  28. Shi, W. et al. Regulation of the pluripotency marker Rex-1 by Nanog and Sox2. J. Biol. Chem. 281, 23319–23325 (2006).

    Article  CAS  Google Scholar 

  29. Cheng, T. et al. Pyruvate carboxylase is required for glutamine-independent growth of tumor cells. Proc. Natl Acad. Sci. USA 108, 8674–8679 (2011).

    Article  CAS  Google Scholar 

  30. Faddah, D. A. et al. Single-cell analysis reveals that expression of nanog is biallelic and equally variable as that of other pluripotency factors in mouse ESCs. Cell Stem Cell 13, 23–29 (2013).

    Article  CAS  Google Scholar 

  31. Boroviak, T., Loos, R., Bertone, P., Smith, A. & Nichols, J. The ability of inner-cell-mass cells to self-renew as embryonic stem cells is acquired following epiblast specification. Nat. Cell Biol. 16, 516–528 (2014).

    Article  CAS  Google Scholar 

  32. Karwacki-Neisius, V. et al. Reduced Oct4 expression directs a robust pluripotent state with distinct signaling activity and increased enhancer occupancy by Oct4 and Nanog. Cell Stem Cell 12, 531–545 (2013).

    Article  CAS  Google Scholar 

  33. Silva, J. et al. Nanog is the gateway to the pluripotent ground state. Cell 138, 722–737 (2009).

    Article  CAS  Google Scholar 

  34. Nichols, J. et al. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell 95, 379–391 (1998).

    Article  CAS  Google Scholar 

  35. Hochedlinger, K. & Jaenisch, R. Induced pluripotency and epigenetic reprogramming. Cold Spring Harb. Perspect. Biol. 7, a019448 (2015).

    Article  Google Scholar 

  36. Silva, J. et al. Promotion of reprogramming to ground state pluripotency by signal inhibition. PLoS Biol. 6, e253 (2008).

    Article  Google Scholar 

  37. Stadtfeld, M., Maherali, N., Borkent, M. & Hochedlinger, K. A reprogrammable mouse strain from gene-targeted embryonic stem cells. Nat. Methods 7, 53–55 (2010).

    Article  CAS  Google Scholar 

  38. Dunn, S. J., Martello, G., Yordanov, B., Emmott, S. & Smith, A. G. Defining an essential transcription factor program for naïve pluripotency. Science 344, 1156–1160 (2014).

    Article  CAS  Google Scholar 

  39. Buganim, Y. et al. The developmental potential of iPSCs is greatly influenced by reprogramming factor selection. Cell Stem Cell 15, 295–309 (2014).

    Article  CAS  Google Scholar 

  40. Weinberger, L., Ayyash, M., Novershtern, N. & Hanna, J. H. Dynamic stem cell states: naive to primed pluripotency in rodents and humans. Nat. Rev. Mol. Cell Biol. 17, 155–169 (2016).

    Article  CAS  Google Scholar 

  41. Choi, J. et al. Prolonged Mek1/2 suppression impairs the developmental potential of embryonic stem cells. Nature 548, 219–223 (2017).

    Article  CAS  Google Scholar 

  42. Yagi, M. et al. Derivation of ground-state female ES cells maintaining gamete-derived DNA methylation. Nature 548, 224–227 (2017).

    Article  CAS  Google Scholar 

  43. Huang, K., Maruyama, T. & Fan, G. The naive state of human pluripotent stem cells: a synthesis of stem cell and preimplantation embryo transcriptome analyses. Cell Stem Cell 15, 410–415 (2014).

    Article  CAS  Google Scholar 

  44. Wang, J. et al. Isolation and cultivation of naive-like human pluripotent stem cells based on HERVH expression. Nat. Protoc. 11, 327–346 (2016).

    Article  CAS  Google Scholar 

  45. Warrier, S. et al. Direct comparison of distinct naive pluripotent states in human embryonic stem cells. Nat. Commun. 8, 15055 (2017).

    Article  CAS  Google Scholar 

  46. Carbognin, E., Betto, R. M., Soriano, M. E., Smith, A. G. & Martello, G. Stat3 promotes mitochondrial transcription and oxidative respiration during maintenance and induction of naive pluripotency. EMBO J. 35, 618–634 (2016).

    Article  CAS  Google Scholar 

  47. Ficz, G. et al. FGF signaling inhibition in ESCs drives rapid genome-wide demethylation to the epigenetic ground state of pluripotency. Cell Stem Cell 13, 351–359 (2013).

    Article  CAS  Google Scholar 

  48. Habibi, E. et al. Whole-genome bisulfite sequencing of two distinct interconvertible DNA methylomes of mouse embryonic stem cells. Cell Stem Cell 13, 360–369 (2013).

    Article  CAS  Google Scholar 

  49. Galonska, C., Ziller, M. J., Karnik, R. & Meissner, A. Ground state conditions induce rapid reorganization of core pluripotency factor binding before global epigenetic reprogramming. Cell Stem Cell 17, 462–470 (2015).

    Article  CAS  Google Scholar 

  50. Bauer, D. E. et al. Cytokine stimulation of aerobic glycolysis in hematopoietic cells exceeds proliferative demand. FASEB J. 18, 1303–1305 (2004).

    Article  CAS  Google Scholar 

  51. Frauwirth, K. A. et al. The CD28 signaling pathway regulates glucose metabolism. Immunity 16, 769–777 (2002).

    Article  CAS  Google Scholar 

  52. Utsunomiya-Tate, N., Endou, H. & Kanai, Y. Cloning and functional characterization of a system ASC-like Na+-dependent neutral amino acid transporter. J. Biol. Chem. 271, 14883–14890 (1996).

    Article  CAS  Google Scholar 

  53. Kammen, H. O. & Hurlbert, R. B. Amination of uridine nucleotides to cytidine nucleotides by soluble mammalian enzymes; role of glutamine and guanosine nucleotides. Biochim. Biophys. Acta 30, 195–196 (1958).

    Article  CAS  Google Scholar 

  54. Marsboom, G. et al. Glutamine metabolism regulates the pluripotency transcription factor OCT4. Cell Rep. 16, 323–332 (2016).

    Article  CAS  Google Scholar 

  55. Alexander, P. B., Wang, J. & McKnight, S. L. Targeted killing of a mammalian cell based upon its specialized metabolic state. Proc. Natl Acad. Sci. USA 108, 15828–15833 (2011).

    Article  CAS  Google Scholar 

  56. Finley, L. W. S. et al. Pluripotency transcription factors and Tet1/2 maintain Brd4-independent stem cell identity. Nat. Cell Biol. 20, 565–574 (2018).

    Article  CAS  Google Scholar 

  57. Shi, Z. D. et al. Genome editing in hPSCs reveals GATA6 haploinsufficiency and a genetic interaction with GATA4 in human pancreatic development. Cell Stem Cell 20, 675–688.e6 (2017).

    Article  CAS  Google Scholar 

  58. Millard, P., Letisse, F., Sokol, S. & Portais, J. C. IsoCor: correcting MS data in isotope labeling experiments. Bioinformatics 28, 1294–1296 (2012).

    Article  CAS  Google Scholar 

  59. Lengner, C. J. et al. Oct4 expression is not required for mouse somatic stem cell self-renewal. Cell Stem Cell 1, 403–415 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the Finley laboratory for discussion and A. Intlekofer for critical feedback. We thank A. Smith (University of Cambridge) for the gift of the chimeric LIF receptor and R. Jaenisch (Whitehead Institute for Biomedical Research) for the gift of the Nanog-GFP ESCs. S.A.V. is a Parker Fellow with the Parker Institute of Cancer Immunotherapy. B.P.R. was supported by an National Institutes of Health (NIH) T32 Training Grant in Molecular and Cellular Biology (no. T32GM008539). L.W.S.F. is a Searle Scholar and was a Dale F. Frey-William Raveis Charitable Fund Scientist supported by the Damon Runyon Cancer Research Foundation (no. DFS-23-17). This work was additionally supported by the Concern Foundation and the Anna Fuller Fund (to L.W.S.F.), The Starr Foundation (no. I11-0039 to L.W.S.F.), a Pathway to Independence Award from the NIH (no. R00 CA191021 to C.C.-F.), NIH/National Institute of Diabetes and Digestive and Kidney Diseases (no. R01DK096239 to D.H.) and the Memorial Sloan Kettering Cancer Center Support Grant no. P30 CA008748.

Author information

Authors and Affiliations

Authors

Contributions

S.A.V. and L.W.S.F. conceived the study. S.A.V., P.K.A and L.W.S.F. performed all the experiments with assistance from Y.C. B.W.C. assisted with the reprogramming experiments. B.P.R. and D.H. performed the human ESC experiments. C.C.-F. performed the immunofluorescence and image analysis. C.B.T. provided additional work in study conception and guidance. S.A.V. and L.W.S.F. wrote the manuscript.

Corresponding author

Correspondence to Lydia W. S. Finley.

Ethics declarations

Competing interests

C.B.T. is a founder of Agios Pharmaceuticals and a member of its scientific advisory board. He also previously served on the board of directors of Merck and Charles River Laboratories.

Additional information

Peer review information: Primary Handling Editor: Ana Mateus

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Figs. 1–6

Reporting Summary

Supplementary Table 1

Source data for all gas chromatography–mass spectrometry experiments

Supplementary Table 2

Calculation of reprogramming efficiency, related to Fig. 4c

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vardhana, S.A., Arnold, P.K., Rosen, B.P. et al. Glutamine independence is a selectable feature of pluripotent stem cells. Nat Metab 1, 676–687 (2019). https://doi.org/10.1038/s42255-019-0082-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-019-0082-3

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing